Articles in PresS. Am J Physiol Lung Cell Mol Physiol (January 30, 2015). doi:10.1152/ajplung.00286.2014 Manuscript for the American Journal of Physiology - Lung Cellular and Molecular Physiology

1

Pericytes contribute to airway remodeling in a mouse model of chronic allergic

2

asthma

3 4

Jill R. Johnson1, 2, *, Erika Folestad1, Jessica E. Rowley2, Elisa M. Noll2, Simone A.

5

Walker2, Clare M. Lloyd2, Sara M. Rankin2, Kristian Pietras1, 3, Ulf Eriksson1 and

6

Jonas Fuxe1

7 8

1

9

Vascular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden

Department of Medical Biochemistry and Biophysics, Matrix Division, Division of

10

2

11

Building, Imperial College London, London SW7 2AZ, United Kingdom

12

3

Leukocyte Biology Section, National Heart and Lung Institute, Sir Alexander Fleming

Lund University, Department of Laboratory Medicine Lund, SE-223 81 Lund, Sweden

13 14

*Corresponding author: Jill Johnson, PhD, Leukocyte Biology Section, National Heart

15

and Lung Institute, Sir Alexander Fleming Building, Imperial College London, London

16

SW7

17

[email protected]

2AZ,

United

Kingdom.

Phone:

+44

(0)

791-448-5692.

18

Copyright © 2015 by the American Physiological Society.

Email:

Johnson et al.

Pericytes in airway remodeling

19

Abstract

20

Myofibroblast accumulation, subepithelial fibrosis and vascular remodeling are

21

complicating features of chronic asthma, but the mechanisms are not clear. Platelet-

22

derived growth factors (PDGFs) regulate the fate and function of various mesenchymal

23

cells and have been implicated as mediators of lung fibrosis. However, it is not known

24

whether PDGF-BB signaling via PDGFR, which is critical for the recruitment of

25

pericytes to blood vessels, plays a role in airway remodeling in chronic asthma.

26

In the present study, we used a selective PDGFRβ inhibitor (CP-673,451) to

27

investigate the role of PDGFR signaling in the development of airway remodeling and

28

lung dysfunction in an established mouse model of house dust mite (HDM) induced

29

chronic allergic asthma.

30

Unexpectedly, we found that pharmacological inhibition of PDGFRβ signaling in

31

the context of chronic aeroallergen exposure led to exacerbated lung dysfunction and

32

airway smooth muscle thickening. Further studies revealed that the inflammatory

33

response to aeroallergen challenge in mice was associated with decreased PDGF-BB

34

expression and the loss of pericytes from the airway microvasculature. In parallel, cells

35

positive for pericyte markers accumulated in the subepithelial region of chronically

36

inflamed airways. This process was exacerbated in animals treated with CP-673,451.

37

The results indicate that perturbed PDGF-BB/PDGFR signaling and pericyte

38

accumulation in the airway wall may contribute to airway remodeling in chronic allergic

39

asthma.

40

Keywords: asthma, house dust mite, airway hyperresponsiveness, pericyte, remodeling

2

Johnson et al.

41

Pericytes in airway remodeling

Introduction

42

Asthma is a heterogenic disease that currently affects 300 million people

43

worldwide (34). Allergic asthma is defined as an allergen-induced, immune-driven

44

chronic lung disease manifested by recurrent episodes of airway inflammation

45

composed of infiltrating eosinophils, mast cells, macrophages, neutrophils and

46

lymphocytes (1). Airway remodeling is thought to be facilitated by this chronic

47

inflammatory process (5). The structural changes associated with airway remodeling

48

comprise goblet cell hyperplasia, collagen deposition and increased airway smooth

49

muscle (ASM) mass, all of which contribute to the clinical manifestation of lung

50

dysfunction (1). Inflammatory cytokines and the pro-fibrogenic growth factor

51

transforming growth factor-β (TGF-β) are believed to play significant roles in driving

52

these structural changes. However, the signaling mechanisms and the cellular sources

53

from which the increased mesenchymal cell population of the airway wall is derived

54

have not been fully described. Some studies have demonstrated that bone marrow-

55

derived fibrocytes contribute to airway wall remodeling (30, 31), while others have

56

suggested a minimal role for these cells (24). Additionally, the differentiation of

57

mesenchymal cells from airway epithelial cells via epithelial-to-mesenchymal transition

58

has been shown to be a mechanism of remodeling in a mouse model of severe allergic

59

airways disease (20).

60

The platelet-derived growth factors (PDGFs) are mitogens for a number of

61

mesenchymal cell types, including fibroblasts and smooth muscle cells (9). The

62

receptors of the PDGFs, PDGF receptor alpha (PDGFRα) and PDGF receptor beta

63

(PDGFRβ) are tyrosine kinases, and are thus amenable to pharmacological

3

Johnson et al.

Pericytes in airway remodeling

64

intervention. However, little is known regarding the expression of the PDGFs in asthma,

65

as the limited studies available in the literature have documented few differences in

66

PDGF or PDGF receptor expression in human asthmatics compared to healthy controls

67

(3, 6, 15, 25). Specifically focusing on PDGF-BB and its cognate receptor PDGFRβ in

68

the context of allergic airway disease, stimulation of ASM cells with PDGF-BB in vitro

69

has been shown to act in concert with TGF-β to stimulate cell migration (18). Moreover,

70

a recent study using an adenovirus vector to overexpress PDGF-BB in the airway

71

epithelium in an OVA-driven mouse model of asthma was shown to induce ASM cell

72

proliferation and enhance airway hyperresponsiveness (14). PDGF receptors are also

73

expressed on vascular mural cells, a heterogeneous population of mesenchymal cells

74

that line the outer surface of microvessels and are therefore abundant in the lung (2).

75

Pericytes, the population of mural cells covering capillaries, express desmin and NG2

76

but are negative for α-smooth muscle actin (α-SMA), while mural cells covering

77

arterioles and venules express desmin and α-SMA and are termed vascular smooth

78

muscle (VSM) cells (2). Mural cells are recruited to and retained on blood vessels

79

through PDGF-BB/PDGFRβ interactions (reviewed in (2)). Impaired pericyte coverage

80

of blood vessels is seen in PDGF-BB-deficient mice and in diseases like cancer, and is

81

associated with vascular leakage and edema (2, 4, 32).

82

In light of these findings, and since tyrosine kinase inhibitors such as masitinib

83

are currently being investigated as asthma therapies (16), we elected to investigate the

84

impact of PDGFRβ inhibition on airway and VSM cells/pericytes in a mouse model of

85

chronic aeroallergen exposure driven by exposure to house dust mite (HDM) extract via

86

the respiratory mucosa. HDM exposure is strongly associated with human asthma and

4

Johnson et al.

Pericytes in airway remodeling

87

is one of the most ubiquitous respiratory allergens worldwide. In mice, chronic

88

respiratory HDM exposure leads to Th2-polarized airway inflammation, remodeling of

89

the airway wall and bronchial hyperreactivity, and thus recapitulates many of the

90

features of clinical asthma (21). Using this paradigm, we investigated the role of

91

PDGFRβ signaling and the downstream effects of inhibiting this receptor during chronic

92

HDM exposure on airway remodeling and lung dysfunction.

93 94

Materials and methods

95

Animal handling

96

Female C57Bl/6 mice were bred in-house at the Karolinska Institutet animal facility at

97

the Department of Mikrobiologi, Tumör- och Cellbiologi (MTC) or purchased from Harlan

98

Laboratories (Wyton, UK) and housed at the central animal facility at Imperial College

99

London. Transgenic mice used for pericyte lineage tracing studies (Tg(Cspg4-

100

DsRed.T1)1Akik/J) were obtained from the Jackson Laboratories (Bar Harbor, ME); the

101

phenotype of these mice was determined by direct fluorescent imaging of the DsRed

102

fluorescent signal in ear biopsies. Animals were initiated into experiments at 8-12 weeks

103

of age. Mice were housed under specific pathogen-free conditions following a 12-h light-

104

dark cycle and were provided food and water ad libitum. All experiments described in

105

this study were approved by the Research Ethics Committees at the Karolinska Institute

106

and at Imperial College London and were performed in accordance with the UK Home

107

Office and Imperial College London guidelines on animal welfare.

108 109

Antigen and drug administration 5

Johnson et al.

Pericytes in airway remodeling

110

Mice were challenged with purified Dermatophagoides pteronyssinus extract (HDM)

111

(Greer Laboratories, Lenoir, NC, USA) 5 days per week for 3 or 5 consecutive weeks.

112

HDM was re-suspended in sterile phosphate buffered saline (PBS) (2.5 mg of

113

protein/ml); 10 μl of HDM were administered intranasally to isoflurane-anesthetized

114

mice (Sigma-Aldrich, Gillingham, UK) (21). Control mice were subjected to the same

115

exposure protocol but received PBS (10 μl). In each group, a subset of animals received

116

a selective PDGFRβ inhibitor (CP-673,451 (Pfizer Inc., New York, NY, USA)) by oral

117

gavage (33 mg/kg in polyethyleneglycol 400 (PEG-400; Sigma-Aldrich) daily for a period

118

of 5 consecutive weeks. Other animals received the drug vehicle (PEG-400). 1-(2-(5-(2-

119

methoxy-ethoxy)-benzoimidazol-1-yl)-quinolin-8-yl)-piperdin-4-ylamine

120

has 10-fold higher affinity for PDGFR versus PDGFR, and greater than 450-fold

121

higher affinity for PDGFR versus c-kit, and was prepared according to the procedures

122

described previously (27).

(CP-673,451),

123 124

Lung function measurements

125

After 5 weeks of allergen and drug administration, mice were subjected to lung function

126

measurements (8). Mice were injected intraperitoneally with sodium pentobarbital (50

127

mg/kg) (Sigma-Aldrich), followed by an intramuscular injection of ketamine (90 mg/kg)

128

(Sigma-Aldrich) (29). Mice were tracheostomized using a 19-gauge blunt-ended needle.

129

Measurements of dynamic airway hyperresponsiveness (AHR) and elastance were

130

acquired using the flexiVent system (Scireq, Montréal, Canada). Mice were ventilated,

131

and airway resistance and elastance were calculated as previously described (29). A 4-

132

6 µm nebulizer was used, and the duration of nebulization was 10 seconds per dose. A

6

Johnson et al.

Pericytes in airway remodeling

133

50% duty cycle was used during nebulization. Fluctuations in AHR and elastance were

134

determined by the responses of the total respiratory system to increasing

135

concentrations (10-300 mg/ml) of methacholine (MCh) (Sigma-Aldrich) delivered into

136

the inspiratory line of the flexiVent ventilator. The PaO waveform was monitored

137

throughout flexiVent data acquisition and was found to be similar during measurements

138

as when the initial calibration was performed before running each subject.

139

Measurements were not taken when there were obvious signs of respiratory effort.

140

Following flexiVent data acquisition, the thoracic cavity of each mouse was opened to

141

confirm that cardiac arrest had not occurred.

142 143

Collection of specimens

144

Mice were humanely sacrificed and the lungs were dissected. In some experiments,

145

bronchoalveolar lavage (BAL) was collected (0.45 ml of PBS). BAL cells were counted

146

using a hemocytometer, centrifuged at 1200 rpm for 5 minutes, then mounted on glass

147

slides using a Cytospin (Thermo Scientific, Hemel Hempstead, UK) and stained with

148

hematoxylin and eosin for differential cell counts. BAL supernatants were stored at -

149

20ºC, then submitted to ELISA to determine mouse PDGF-BB expression according to

150

the manufacturer’s instructions (R&D Systems, Abingdon, UK). In other experiments,

151

mice were perfused through the left ventricle with 1% paraformaldehyde, and the

152

trachea, extrapulmonary bronchi and lung were dissected out. The tracheas,

153

extrapulmonary

154

paraformaldehyde for 30 min, then transferred to PBS. The lung was embedded in

bronchi

and

lungs

were

isolated

and

preserved

in

1%

7

Johnson et al.

Pericytes in airway remodeling

155

OCT, stored at -80°C and cut into 10 μm thick sections for staining. Fresh lung samples

156

were snap-frozen for qPCR and immunoblotting experiments.

157 158

Immunoblotting

159

Frozen lung tissue specimens were homogenized in 400 μl of lysis buffer, which was

160

composed of T-PER Tissue Protein Extraction Reagent (Rockford, IL) supplemented

161

with protease inhibitors (Complete Mini tablets from Roche Diagnostics Scandinavia,

162

Bromma, Sweden), and homogenized with a homogenizer (Percell, Stockholm,

163

Sweden). Protein samples were separated by SDS-PAGE on 4-12% polyacrylamide

164

gels and blotted onto nitrocellulose membranes. The membranes were incubated in

165

blocking buffer (5% skim milk) in Tris-buffered saline containing 0.5% Tween for 1 hour

166

at room temperature and probed with a polyclonal rabbit-antihuman PDGF-BB antibody

167

(Aviva Systems Biology, San Diego, CA) diluted 1:1000 in blocking buffer. After

168

washing, the membranes were incubated with a HRP-conjugated secondary anti-rabbit

169

antibody (GE Healthcare, Stockholm, Sweden) diluted 1:5000 in blocking buffer, for 1

170

hour at RT. The membranes were developed using ECL+ detection reagent

171

(Amersham, GE Healthcare). To ascertain equal loading of protein, the membranes

172

were stripped and re-probed with a rabbit-anti-mouse antibody against Akt (Cell

173

Signaling/BioNordika Sweden, Stockholm, Sweden) diluted 1:1000 in blocking buffer.

174 175

Immunofluorescent staining

176

Lung sections from C57Bl6 and DsRed-NG2 mice were warmed to room temperature

177

and non-specific binding was blocked by incubating in 5% normal goat serum (NGS),

8

Johnson et al.

Pericytes in airway remodeling

178

0.3% Triton-X 100 and 0.1% bovine serum albumin (BSA) (all purchased from Sigma-

179

Aldrich) in PBS for 1 h at RT. Sections were stained using the appropriate primary (O/N)

180

and secondary (1 h) antibodies (Table 1). After washing twice in PBS, sections were

181

mounted using a glycerol-based mounting medium containing DAPI (Vector

182

Laboratories, Peterborough, UK) to visualize DNA. For polyclonal antibodies, negative

183

reagent controls were carried out.

184

For

tracheobronchal

whole

mount

immunostaining,

the

trachea

and

185

extrapulmonary bronchi were dissected and cleaned of connective tissue and mounted

186

on KwikgardTM-coated (WPI, Sarasota, FL, USA) 6-well culture plates. Non-specific

187

binding was blocked by incubating trachea sections in 5% NGS, 0.3% Triton-X 100 and

188

0.2% BSA in PBS O/N. Left and right trachea sections were stained using the

189

appropriate primary and secondary antibodies O/N at RT (Table 1). Sections were

190

washed thrice in PBS and mounted using a glycerol-based mounting medium containing

191

DAPI (Vector Laboratories). For all antibodies, negative reagent controls were carried

192

out with no staining observed.

193 194

Light microscopy

195

Quantification of αSMA and NG2 positive cells in the trachea and extrapulmonary

196

bronchi was performed using a Leica DM2500 fluorescence microscope (Leica

197

Microsystems, Milton Keynes, UK). Counts were normalized to the length of each

198

tracheobronchal whole mount (including the extrapulmonary bronchi at the distal end of

199

the preparation). For image acquisition from immunostained tracheobronchal whole

200

mounts and lung sections, 1024 x 1024 pixel RGB-color images were obtained using a

9

Johnson et al.

Pericytes in airway remodeling

201

Zeiss LSM 510 inverted confocal microscope with argon, helium-neon and UV lasers

202

(Carl Zeiss AG, Göttingen, Germany) using an EC Plan-Neofluar 40x/1.30 oil objective

203

with the DIC phase contrast technique (10). Using the optimized pinhole size, 0.9 μm

204

thick optical sections were sequentially collected with a step size of 1 μm (lung sections)

205

or 1.5 μm (tracheas). Image analysis was performed using Zeiss AIM 3.2.2 software

206

and Zeiss LSM Image software (Carl Zeiss AG). Three-dimensional visualizations

207

(Videos 1-4) of confocal images were prepared using Volocity software (Perkin-Elmer,

208

Waltham, MA). Morphometric analysis of ASM thickness was performed by a blinded

209

observer on αSMA-stained lung sections and analyzed using ImageJ software (NIH,

210

USA). Morphometric analysis of the number of NG2+ cells in the airway subepithelial

211

region was conducted by a blinded observer on lung sections stained for NG2, SMA

212

and CD31 (to prevent enumeration of pericytes associated with the microvasculature).

213

Five representative images were taken of the large airways (bronchi and bronchioles)

214

from each animal (n=5-10 per group from two independent experiments). Images were

215

taken on a Zeiss LSM 510 inverted confocal microscope and processed using ImageJ.

216

NG2+ cells in the airway subepithelial region were enumerated and counts were

217

normalized to mm of basement membrane.

218 219

Quantitative PCR (qPCR)

220

Whole tissue RNA from lung was isolated with TRIzol reagent (Invitrogen/Life

221

Technologies, Stockholm, Sweden) and QIAGEN RNeasy (Qiagen, Sollentuna,

222

Sweden) according to the manufacturers’ instructions. Total RNA (1 μg) was reverse

223

transcribed according to the manufacturer’s instructions (iScript cDNA synthesis kit, Bio-

10

Johnson et al.

Pericytes in airway remodeling

224

Rad). qPCR was performed using Platinum SYBR green SuperMix (Invitrogen) and

225

25 ng cDNA per reaction. Primers used were: Pdgfb QT00266910, Pdgfrb QT00113148,

226

L19 QT01779218. Ct values for the three genes analyzed were: 19-20 (PDGF-BB); 24-

227

26 (PDGFRβ); 17-18 (L19). Expression levels were normalized to the expression of

228

L19. Melt curves were assessed following the completion of RT-PCR to ensure that the

229

fluorescent signal resulted from a single PCR product.

230 231

Data analysis

232

Data were analyzed with GraphPad Prism Software version 6 (GraphPad Software, San

233

Diego, CA, USA). Statistical analysis was performed by Student’s t-test or one-way

234

analysis of variance (ANOVA) with a subsequent Tukey post-hoc test where

235

appropriate. Differences were considered statistically significant when p

Pericytes contribute to airway remodeling in a mouse model of chronic allergic asthma.

Myofibroblast accumulation, subepithelial fibrosis, and vascular remodeling are complicating features of chronic asthma, but the mechanisms are not cl...
2MB Sizes 0 Downloads 8 Views